Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cells ; 8(12)2019 11 28.
Article in English | MEDLINE | ID: mdl-31795206

ABSTRACT

Cardiac Progenitor Cells (CPCs) show great potential as a cell resource for restoring cardiac function in patients affected by heart disease or heart failure. CPCs are proliferative and committed to cardiac fate, capable of generating cells of all the cardiac lineages. These cells offer a significant shift in paradigm over the use of human induced pluripotent stem cell (iPSC)-derived cardiomyocytes owing to the latter's inability to recapitulate mature features of a native myocardium, limiting their translational applications. The iPSCs and direct reprogramming of somatic cells have been attempted to produce CPCs and, in this process, a variety of chemical and/or genetic factors have been evaluated for their ability to generate, expand, and maintain CPCs in vitro. However, the precise stoichiometry and spatiotemporal activity of these factors and the genetic interplay during embryonic CPC development remain challenging to reproduce in culture, in terms of efficiency, numbers, and translational potential. Recent advances in biomaterials to mimic the native cardiac microenvironment have shown promise to influence CPC regenerative functions, while being capable of integrating with host tissue. This review highlights recent developments and limitations in the generation and use of CPCs from stem cells, and the trends that influence the direction of research to promote better application of CPCs.


Subject(s)
Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Myoblasts, Cardiac/cytology , Myoblasts, Cardiac/metabolism , Animals , Biocompatible Materials , Cell Differentiation/genetics , Cells, Cultured , Cellular Reprogramming/genetics , Cellular Reprogramming Techniques , Genetic Engineering , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Regeneration , Stem Cell Transplantation , Tissue Engineering
2.
Stem Cells Dev ; 26(23): 1695-1705, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28992755

ABSTRACT

Cardiomyocytes (CMs) derived from human induced pluripotent stem cells (hiPSCs) are now a well-established modality for modeling genetic disorders of the heart. This is especially so for long QT syndrome (LQTS), which is caused by perturbation of ion channel function, and can lead to fainting, malignant arrhythmias and sudden cardiac death. LQTS2 is caused by mutations in KCNH2, a gene whose protein product contributes to IKr (also known as HERG), which is the predominant repolarizing potassium current in CMs. ß-blockers are the mainstay treatment for patients with LQTS, functioning by reducing heart rate and arrhythmogenesis. However, they are not effective in around a quarter of LQTS2 patients, in part, because they do not correct the defining feature of the condition, which is excessively prolonged QT interval. Since new therapeutics are needed, in this report, we biopsied skin fibroblasts from a patient who was both genetically and clinically diagnosed with LQTS2. By producing LQTS-hiPSC-CMs, we assessed the impact of different drugs on action potential duration (APD), which is used as an in vitro surrogate for QT interval. Not surprisingly, the patient's own ß-blocker medication, propranolol, had a marginal effect on APD in the LQTS-hiPSC-CMs. However, APD could be significantly reduced by up to 19% with compounds that enhanced the IKr current by direct channel binding or by indirect mediation through the PPARδ/protein 14-3-3 epsilon/HERG pathway. Drug-induced enhancement of an alternative potassium current, IKATP, also reduced APD by up to 21%. This study demonstrates the utility of LQTS-hiPSC-CMs in evaluating whether drugs can shorten APD and, importantly, shows that PPARδ agonists may form a new class of therapeutics for this condition.


Subject(s)
Action Potentials , ERG1 Potassium Channel/metabolism , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Adrenergic beta-Antagonists/pharmacology , Cell Differentiation , Cells, Cultured , ERG1 Potassium Channel/genetics , Female , Fibroblasts/cytology , Humans , Long QT Syndrome/genetics , Long QT Syndrome/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Propranolol/pharmacology , Young Adult
3.
Stem Cells Dev ; 25(6): 439-52, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26906236

ABSTRACT

Automated planar patch clamp systems are widely used in drug evaluation studies because of their ability to provide accurate, reliable, and reproducible data in a high-throughput manner. Typically, CHO and HEK tumorigenic cell lines overexpressing single ion channels are used since they can be harvested as high-density, homogenous, single-cell suspensions. While human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) are physiologically more relevant, these cells are fragile, have complex culture requirements, are inherently heterogeneous, and are expensive to produce, which has restricted their use on automated patch clamp (APC) devices. Here, we used high efficiency differentiation protocols to produce cardiomyocytes from six different hPSC lines for analysis on the Patchliner (Nanion Technologies GmbH) APC platform. We developed a two-step cell preparation protocol that yielded cell catch rates and whole-cell breakthroughs of ∼80%, with ∼40% of these cells allowing electrical activity to be recorded. The protocol permitted formation of long-lasting (>15 min), high quality seals (>2 GΩ) in both voltage- and current-clamp modes. This enabled density of sodium, calcium, and potassium currents to be evaluated, along with dose-response curves to their respective channel inhibitors, tetrodotoxin, nifedipine, and E-4031. Thus, we show the feasibility of using the Patchliner platform for automated evaluation of the electrophysiology and pharmacology of hPSC-CMs, which will enable considerable increase in throughput for reliable and efficient drug evaluation.


Subject(s)
High-Throughput Screening Assays/methods , Myocytes, Cardiac/cytology , Patch-Clamp Techniques/methods , Pluripotent Stem Cells/cytology , Primary Cell Culture/methods , Action Potentials , Calcium/metabolism , Cell Differentiation , Cells, Cultured , Humans , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Pluripotent Stem Cells/physiology , Potassium/metabolism , Potassium Channel Blockers/pharmacology , Sodium/metabolism , Sodium Channel Blockers/pharmacology
4.
Biochim Biophys Acta ; 1863(7 Pt B): 1728-48, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26524115

ABSTRACT

Cardiomyocytes from human pluripotent stem cells (hPSCs-CMs) could revolutionise biomedicine. Global burden of heart failure will soon reach USD $90bn, while unexpected cardiotoxicity underlies 28% of drug withdrawals. Advances in hPSC isolation, Cas9/CRISPR genome engineering and hPSC-CM differentiation have improved patient care, progressed drugs to clinic and opened a new era in safety pharmacology. Nevertheless, predictive cardiotoxicity using hPSC-CMs contrasts from failure to almost total success. Since this likely relates to cell immaturity, efforts are underway to use biochemical and biophysical cues to improve many of the ~30 structural and functional properties of hPSC-CMs towards those seen in adult CMs. Other developments needed for widespread hPSC-CM utility include subtype specification, cost reduction of large scale differentiation and elimination of the phenotyping bottleneck. This review will consider these factors in the evolution of hPSC-CM technologies, as well as their integration into high content industrial platforms that assess structure, mitochondrial function, electrophysiology, calcium transients and contractility. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.


Subject(s)
Biomedical Research/methods , Cardiovascular Agents/pharmacology , Cell Lineage , Drug Discovery/methods , Heart Diseases/drug therapy , High-Throughput Screening Assays , Induced Pluripotent Stem Cells/physiology , Myocytes, Cardiac/physiology , Toxicity Tests/methods , Cardiovascular Agents/toxicity , Cell Differentiation , Cell Proliferation , Cells, Cultured , Genotype , Heart Diseases/chemically induced , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Diseases/physiopathology , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Phenotype , Risk Assessment
5.
Adv Mater ; 27(27): 4006-12, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26033422

ABSTRACT

A scalable and cost-effective synthetic polymer substrate that supports robust expansion and subsequent multilineage differentiation of human pluripotent stem cells (hPSCs) with defined commercial media is presented. This substrate can be applied to common cultureware and used off-the-shelf after long-term storage. Expansion and differentiation of hPSCs are performed entirely on the polymeric surface, enabling the clinical potential of hPSC-derived cells to be realized.


Subject(s)
Cell Culture Techniques/instrumentation , Cell Differentiation/physiology , Pluripotent Stem Cells/physiology , Polymers , Cell Adhesion/physiology , Cell Line , Cell Lineage , Culture Media , Fluorescent Antibody Technique , High-Throughput Screening Assays , Humans , Microarray Analysis
6.
Stem Cells Dev ; 22(20): 2714-24, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23829870

ABSTRACT

With an incidence of ∼1:3,500 to 5,000 in male children, Duchenne muscular dystrophy (DMD) is an X-linked disorder in which progressive muscle degeneration occurs and affected boys usually die in their twenties or thirties. Cardiac involvement occurs in 90% of patients and heart failure accounts for up to 40% of deaths. To enable new therapeutics such as gene therapy and exon skipping to be tested in human cardiomyocytes, we produced human induced pluripotent stem cells (hiPSC) from seven patients harboring mutations across the DMD gene. Mutations were retained during differentiation and analysis indicated the cardiomyocytes showed a dystrophic gene expression profile. Antisense oligonucleotide-mediated skipping of exon 51 restored dystrophin expression to ∼30% of normal levels in hiPSC-cardiomyocytes carrying exon 47-50 or 48-50 deletions. Alternatively, delivery of a dystrophin minigene to cardiomyocytes with a deletion in exon 35 or a point mutation in exon 70 allowed expression levels similar to those seen in healthy cells. This demonstrates that DMD hiPSC-cardiomyocytes provide a novel tool to evaluate whether new therapeutics can restore dystrophin expression in the heart.


Subject(s)
Dystrophin/genetics , Gene Expression/drug effects , Induced Pluripotent Stem Cells/metabolism , Muscular Dystrophy, Duchenne/genetics , Mutation , Myocytes, Cardiac/drug effects , Oligonucleotides, Antisense/pharmacology , Base Sequence , Cell Differentiation , Child , Dystrophin/metabolism , Exons , Gene Transfer Techniques , Genetic Therapy , Humans , Induced Pluripotent Stem Cells/pathology , Male , Molecular Sequence Data , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/therapy , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Primary Cell Culture
7.
Stem Cells Dev ; 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23786351

ABSTRACT

With an incidence of ~1:3,500 to 5,000 in male children, Duchenne muscular dystrophy (DMD) is an X-linked disorder in which progressive muscle degeneration occurs and affected boys usually die in their twenties or thirties. Cardiac involvement occurs in 90% of patients and heart failure accounts for up to 40% of deaths. To enable new therapeutics such as gene therapy and exon skipping to be tested in human cardiomyocytes, we produced human induced pluripotent stem cells (hiPSC) from seven patients harbouring mutations across the DMD gene. Mutations were retained during differentiation and analysis indicated the cardiomyocytes showed a dystrophic gene expression profile. Antisense oligonucleotide-mediated skipping of exon 51 restored dystrophin expression to ~30% of normal levels in hiPSC-cardiomyocytes carrying exon 47-50 or 48-50 deletions. Alternatively, delivery of a dystrophin minigene to cardiomyocytes with a deletion in exon 35 or a point mutation in exon 70 allowed expression levels similar to those seen in healthy cells. This demonstrates that DMD hiPSC-cardiomyocytes provide a novel tool to evaluate whether new therapeutics can restore dystrophin expression in the heart.

8.
Biochim Biophys Acta ; 1830(6): 3517-24, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23403134

ABSTRACT

BACKGROUND: Online label-free monitoring of in-vitro differentiation of stem cells remains a major challenge in stem cell research. In this paper we report the use of Raman micro-spectroscopy (RMS) to measure time- and spatially-resolved molecular changes in intact embryoid bodies (EBs) during in-vitro cardiogenic differentiation. METHODS: EBs formed by aggregation of human embryonic stem cells (hESCs) were cultured in defined medium to induce differentiation towards cardiac phenotype and maintained in purpose-built micro-bioreactors on the Raman microscope for 5days (between days 5 and 9 of differentiation) and spatially-resolved spectra were recorded at 24h intervals. RESULTS: The Raman spectra showed that the onset of spontaneous beating of EBs at day 7 coincided with an increase in the intensity of the Raman bands at 1340cm(-1), 1083cm(-1), 937cm(-1), 858cm(-1), 577cm(-1) and 482cm(-1). The spectral maps corresponding to these bands had a high positive correlation with the expression of the cardiac-specific α-actinin obtained by immuno-fluorescence imaging of the same EBs. The spectral markers obtained here are also in agreement with previous studies performed on individual live hESC-derived CMs. CONCLUSIONS: The intensity profile of these Raman bands can be used for label-free in-situ monitoring of EBs to estimate the efficacy of cardiogenic differentiation. GENERAL SIGNIFICANCE: As the acquisition of the time-course Raman spectra did not affect the viability or the differentiation potential of the hESCs, this study demonstrates the feasibility of using RMS for on-line non-invasive continuous monitoring of such processes inside bioreactor culture systems.


Subject(s)
Cell Differentiation , Embryonic Stem Cells , Myocytes, Cardiac , Spectrum Analysis, Raman/methods , Actinin/biosynthesis , Antigens, Differentiation/biosynthesis , Cell Culture Techniques , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Humans , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism
9.
Bioessays ; 35(3): 281-98, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22886688

ABSTRACT

The emphasis in human pluripotent stem cell (hPSC) technologies has shifted from cell therapy to in vitro disease modelling and drug screening. This review examines why this shift has occurred, and how current technological limitations might be overcome to fully realise the potential of hPSCs. Details are provided for all disease-specific human induced pluripotent stem cell lines spanning a dozen dysfunctional organ systems. Phenotype and pharmacology have been examined in only 17 of 63 lines, primarily those that model neurological and cardiac conditions. Drug screening is most advanced in hPSC-cardiomyocytes. Responses for almost 60 agents include examples of how careful tests in hPSC-cardiomyocytes have improved on existing in vitro assays, and how these cells have been integrated into high throughput imaging and electrophysiology industrial platforms. Such successes will provide an incentive to overcome bottlenecks in hPSC technology such as improving cell maturity and industrial scalability whilst reducing cost.


Subject(s)
Disease Models, Animal , Drug Evaluation, Preclinical , Pluripotent Stem Cells/metabolism , Animals , High-Throughput Screening Assays , Humans , Phenotype , Pluripotent Stem Cells/cytology , Stem Cell Transplantation
10.
J Biomed Opt ; 16(4): 045002, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21529069

ABSTRACT

Raman micro-spectroscopy (RMS) has been recently proposed for label-free phenotypic identification of human embryonic stem cells (hESC)-derived cardiomyocytes. However, the methods used for measuring the Raman spectra led to acquisition times of minutes per cell, which is prohibitive for rapid cell sorting applications. In this study we evaluated two measurement strategies that could reduce the measurement time by a factor of more than 100. We show that sampling individual cells with a laser beam focused to a line could eliminate the need of cell raster scanning and achieve high prediction accuracies (>95% specificity and >96% sensitivity) with acquisition times ∼ 5 seconds per cell. However, the use of commercially-available higher power lasers could potentially lead to sorting speeds of ∼ 10 cells per s. This would start to progress RMS to the field of cell sorting for applications such as enrichment and purification of hESC-derived cardiomyocytes.


Subject(s)
Cell Separation/methods , Embryonic Stem Cells/cytology , Myocytes, Cardiac/cytology , Spectrum Analysis, Raman/methods , Embryonic Stem Cells/chemistry , Flow Cytometry , Humans , Myocytes, Cardiac/chemistry , Phenotype , Principal Component Analysis , Sensitivity and Specificity , Time Factors
11.
J Pharmacol Exp Ther ; 328(1): 99-106, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18952886

ABSTRACT

Chronic cocaine exposure is associated with severe cardiac complications, but the mechanisms of cocaine cardiotoxicity remain unclear, and current therapies are unsatisfactory. We investigated the hypothesis of oxidative stress-mediated cardiotoxicity and the role of NADPH oxidase in this process in a mouse model of chronic escalating "binge" cocaine administration (milligrams per kilogram): days 1 to 4 at 3 x 15 mg, days 5 to 8 at 3 x 20 mg, days 9 to 12 at 3 x 25 mg, and days 13 to 14 at 3 x 30 mg. Compared with vehicle controls, chronic binge cocaine administration significantly increased the cardiac NADPH-dependent O(2)(.) production (1.96- +/- 0.4-fold) as detected by tiron (an O(2)(.) scavenger)-inhibitable lucigenin chemiluminescence and dihydroethidium fluorescence. Cocaine-induced reactive oxygen species (ROS) production was associated with significant increases ( approximately 2-fold) in the protein expressions of Nox2 (an isoform of NADPH oxidase) and its regulatory subunits: p22(phox), p67(phox), p47(phox), p40(phox), and Rac1, and in p47(phox) phosphorylation as detected by immunoblotting (all p < 0.03). Increased Nox2 activity was accompanied by the activation of extracellular signal-regulated kinase 1/2, p38 mitogen-activated protein kinase (MAPK), and c-Jun NH(2)-terminal kinase, notably in the cardiomyocytes. Cell culture experiments revealed that cocaine-induced ROS production was primarily a direct action of cocaine on cardiac myocytes, which caused severe oxidative damage to myocytes and cell death as detected by terminal deoxynucleotidyl transferase dUTP nick-end labeling assay. These could be inhibited by inhibitors to protein kinase C (bisindolymaleimide) or by depletion of Nox2 using small interfering RNA. In conclusion, chronic cocaine administration directly causes severe myocardial oxidative stress through the activation of Nox2 oxidase. Increased ROS production contributes to MAPK activation and the subsequent myocyte damage. Inhibitors to NADPH oxidase or antioxidants may have therapeutic potential in the treatment of cocaine cardiotoxicity.


Subject(s)
Cocaine/toxicity , Heart/physiopathology , Membrane Glycoproteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , NADPH Oxidases/metabolism , Oxidative Stress/drug effects , Animals , Cell Line , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Heart/drug effects , In Situ Nick-End Labeling , Male , Membrane Glycoproteins/drug effects , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Myocardium/enzymology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , NADPH Oxidase 2 , NADPH Oxidases/drug effects , NADPH Oxidases/genetics , Rats , Reactive Oxygen Species/metabolism , Superoxides/metabolism , Transfection
12.
Mol Biol Cell ; 18(10): 4168-79, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17699598

ABSTRACT

We have investigated the cellular responses to hydrostatic pressure by using the fission yeast Schizosaccharomyces pombe as a model system. Exposure to sublethal levels of hydrostatic pressure resulted in G2 cell cycle delay. This delay resulted from Cdc2 tyrosine-15 (Y-15) phosphorylation, and it was abrogated by simultaneous disruption of the Cdc2 kinase regulators Cdc25 and Wee1. However, cell cycle delay was independent of the DNA damage, cytokinesis, and cell size checkpoints, suggesting a novel mechanism of Cdc2-Y15 phosphorylation in response to hydrostatic pressure. Spc1/Sty1 mitogen-activated protein (MAP) kinase, a conserved member of the eukaryotic stress-activated p38, mitogen-activated protein (MAP) kinase family, was rapidly activated after pressure stress, and it was required for cell cycle recovery under these conditions, in part through promoting polo kinase (Plo1) phosphorylation on serine 402. Moreover, the Spc1 MAP kinase pathway played a key role in maintaining cell viability under hydrostatic pressure stress through the bZip transcription factor, Atf1. Further analysis revealed that prestressing cells with heat increased barotolerance, suggesting adaptational cross-talk between these stress responses. These findings provide new insight into eukaryotic homeostasis after exposure to pressure stress.


Subject(s)
Cell Cycle , Schizosaccharomyces/cytology , Adaptation, Physiological , Cytokinesis , DNA, Fungal/metabolism , Enzyme Activation , G2 Phase , Hydrostatic Pressure , Kinetics , Microbial Viability , Models, Biological , Schizosaccharomyces pombe Proteins/metabolism
13.
Free Radic Biol Med ; 43(6): 976-86, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17697942

ABSTRACT

Endothelial cells (EC) express constitutively two major isoforms (Nox2 and Nox4) of the catalytic subunit of NADPH oxidase, which is a major source of endothelial reactive oxygen species. However, the individual roles of these Noxes in endothelial function remain unclear. We have investigated the role of Nox2 in nutrient deprivation-induced cell cycle arrest and apoptosis. In proliferating human dermal microvascular EC, Nox2 mRNA expression was low relative to Nox4 (Nox2:Nox4 approximately 1:13), but was upregulated 24 h after starvation and increased to 8+/-3.5-fold at 36 h of starvation. Accompanying the upregulation of Nox2, there was a 2.28+/-0.18-fold increase in O2.- production, a dramatic induction of p21cip1 and p53, cell cycle arrest, and the onset of apoptosis (all p<0.05). All these changes were inhibited significantly by in vitro deletion of Nox2 expression and in coronary microvascular EC isolated from Nox2 knockout mice. In Nox2 knockout cells, although there was a 3.8+/-0.5-fold increase in Nox4 mRNA expression after 36 h of starvation (p<0.01), neither O2.- production nor the p21cip1 or p53 expression was increased significantly and only 0.46% of cells were apoptotic. In conclusion, Nox2-derived O2.-, through the modulation of p21cip1 and p53 expression, participates in endothelial cell cycle regulation and apoptosis.


Subject(s)
Apoptosis , Cell Cycle , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Endothelium, Vascular/cytology , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/genetics , Cell Cycle/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA, Antisense/pharmacology , Endothelium, Vascular/enzymology , Humans , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , NADPH Oxidase 1 , NADPH Oxidase 2 , NADPH Oxidase 4 , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , Oxidative Stress , RNA, Messenger/analysis , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...